• Non ci sono risultati.

Impact of Diabetes on Outcomes of Sorafenib Therapy for Hepatocellular Carcinoma

N/A
N/A
Protected

Academic year: 2021

Condividi "Impact of Diabetes on Outcomes of Sorafenib Therapy for Hepatocellular Carcinoma"

Copied!
7
0
0

Testo completo

(1)

ORIGINAL RESEARCH ARTICLE

Impact of Diabetes on Outcomes of Sorafenib Therapy

for Hepatocellular Carcinoma

Giovan Giuseppe Di Costanzo1&Raffaella Tortora1&Filomena Morisco2&

Luigi Addario1&Maria Guarino2&Gabriella Cordone1&Luigia Falco1&

Nicola Caporaso2

Published online: 9 August 2016

# Springer International Publishing Switzerland 2016

Abstract

Background Patients with diabetes are at increased risk of developing hepatocellular carcinoma (HCC) and have a poorer prognosis as compared to non-diabetics when HCC occurs. Diabetics with non-HCC cancers are at higher risk of toxicity related to systemic therapy, but data on HCC are lacking.

Objective The aim of this study was to evaluate safety and effectiveness of sorafenib in HCC patients according to the presence/absence of diabetes.

Patients and Methods From October 2008 to June 2014, 313 patients with HCC treated with sorafenib were enrolled. The patients were staged according to the BCLC system. Treatment response was evaluated according to the mRECIST criteria. The main evaluated outcomes were the overall survival and the safety in the two groups.

Results Patients were divided in two groups: 80 diabetics (DIAB) and 233 nondiabetics (nDIAB). The median treatment duration was 4 months in DIAB and 3 months in nDIAB. Main adverse events occurred with comparable frequency in both groups, with the exception of rash, that was more fre-quent among DIAB than in nDIAB: 27.5 % vs 17.6 % (P = .047). The median overall survival was 9 months in nDIAB and 10 months in DIAB group (P = .535). Median time-to-progression (TTP) was longer the in DIAB than the nDIAB group (P = .038).

Conclusions Sorafenib was as safe as effective in DIAB and in nDIAB patients. The longer TTP observed among DIAB than in nDIAB patients might suggest a better anticancer ef-fect of sorafenib in patients with diabetes.

Key Points

Sorafenib in diabetics was safe and the longer TTP observed in diabetics might suggest a better anticancer effect of sorafenib in patients with diabetes.

The better TTP in patients treated with oral therapy as compared to those receiving insulin is interesting and concordant with previous studies on different cancers.

Abbreviations

HCC Hepatocellular carcinoma

HCV Hepatitis C virus

HBV Hepatitis B virus

EASL European Association for the study of the liver BCLC Barcelona clinic liver cancer

ECOG Eastern cooperative oncology group

PS Performance status

AFP Alpha fetoprotein

US Ultrasound

CT Computed tomography

MRI Magnetic resonance imaging

OS Overall survival

TTP Time-to-progression

mRECIST Modified Response Evaluation Criteria in Solid Tumours for hepatocellular carcinoma

CR Complete response

PR Partial response

* Giovan Giuseppe Di Costanzo

ggdicostanzo@libero.it; dicostanzogg@gmail.com

1

Department of Transplantation - Liver Unit, Cardarelli Hospital, Via A. Cardarelli 9, 80131 Naples, Italy

2 Department of Clinical Medicine and Surgery - Gastroenterology Unit, University of NaplesBFederico II^, Naples, Italy

(2)

SD Stable disease

PD Progressive disease

DIAB Diabetics

nDIAB Nondiabetics

HFS Hand foot syndrome

TACE Transarterial chemoembolization

1 Introduction

Hepatocellular carcinoma (HCC) is a leading cause of death for cancer worldwide [1]. In Western countries, HCC occurs in most of cases in patients with cirrhosis, that is, therefore, a preneoplastic state, and is the main cause of death among cirrhotics [2,3]. Hepatitis C (HCV) and B (HBV) viral infec-tions and alcohol consumption are the main etiological agents. In the last years, nonalcoholic fatty liver disease and steatohepatitis are becoming increasingly recognized causes of cirrhosis and HCC [4]. Metabolic syndrome, mainly obesity and diabetes, is strictly related to the presence of liver steatosis and steatohepatitis [5]. Patients with obesity and type 2 diabetes are at increased risk of developing HCC [6, 7]. A longer duration of diabetes is related to a higher rate of HCC development [8,9], and diabetes may worsen the outcomes of patients with cirrhosis and HCC [10–13]. Furthermore, im-paired carbohydrate metabolism is more frequently observed in cirrhosis than in the general population and about a third of these patients have type 2 diabetes [14].

In spite of more extensive use of surveillance and liver imaging in patients with cirrhosis, a substantial proportion of HCC are diagnosed in intermediate and advanced stage and only 30 % of patients can be treated with potentially curative therapies. Moreover, over time a considerable percentage of early HCC patients progresses to the advanced stage [15]. Therefore, many patients with HCC necessitate systemic treat-ment, and sorafenib (Nexavar©, Bayer Health Care, Leverkusen, Germany) is the only drug approved worldwide for HCC advanced stage patients who are not candidates to curative treatments [16]. However, systemic therapy in pa-tients with diabetes might be challenging. Papa-tients with diabe-tes and non-HCC cancers are at higher risk of toxicity related to systemic treatment and often are treated with less aggres-sive therapies as compared to patients without diabetes [17,

18]. Data regarding HCC patients are lacking and only a small study on patients with metastatic renal cell carcinoma or ad-vanced HCC showed that sorafenib treatment in diabetic pa-tients was safe [19]. Due to the high prevalence and the role of diabetes in HCC these data are necessary and clinically rele-vant. Therefore, the aim of this field-practice study was to evaluate safety and effectiveness of sorafenib treatment in a large cohort of HCC patients according to the presence or absence of diabetes.

2 Patients and Methods

2.1 Patients

From October 2008 to June 2014, all consecutive patients with unresectable HCC who were treated with sorafenib at-tending the Liver Unit of Cardarelli Hospital were included in this single centre prospective observational study. The diag-nosis of HCC was performed according to the European Association for the Study of the Liver (EASL) criteria [20]. At the time of starting the treatment with sorafenib, the tumor stage was assessed following the Barcelona Clinic Liver Cancer (BCLC) staging system [21]. Patients were treated with sorafenib in case of HCC untreatable with surgery or locoregional therapy, ECOG (Eastern Cooperative Oncology Group) performance status (PS) 0–1, and Child-Pugh score ≤ 7. Starting dose of sorafenib was 800 mg/day, except for pa-tients with bilirubin >2 mg/dL or ascites in whom the dose was reduced to 400 mg/day.

The diagnosis of diabetes was performed when fasting blood glucose was greater than or equal to 126 mg/dl, accord-ing to the American Diabetes Association guidelines 2015 [22].

2.2 Assessment Schedule

At baseline, all necessary patient characteristics were evaluat-ed: age, sex, height, weight, clinical information, biochemical parameters, serum alpha-fetoprotein (AFP), upper gastrointes-tinal endoscopy, abdominal Doppler ultrasound (US), chest computed tomography (CT), and abdominal contrast-enhanced CT or magnetic resonance imaging (MRI). In pa-tients with suspected bone metastasis, a bone scintigraphy was performed. The treatment duration was calculated by sum-ming the days of drug intake, excluding the intervals of tem-porary discontinuation of sorafenib.

Once a month patients underwent to clinical evaluation, biochemical tests, AFP, abdominal US, pill-count for the eval-uation of compliance to treatment, and record of adverse events according to definitions from the National Cancer Institute's Common Toxicity Criteria version 3.0 (http://ctep. info.nih.gov/CTC3/ctc ind term.html), using a pre-planned questionnaire. For a better management of patients, a call cen-tre was available every day to patients for consulting a doctor. To evaluate radiological response to treatment, CT or MRI scans were performed every 3 months and in any case of suspected progression.

2.3 Outcomes and Assessments

The main evaluated outcomes were the overall survival (OS) and the safety in the two groups.

(3)

The OS was measured from the date of starting sorafenib until the date of death or the last visit. The safety was assessed evaluating the occurrence and the grade of sorafenib adverse events.

Secondary outcomes were the 3-month radiological re-sponse to sorafenib and the time-to-progression (TTP). The radiological response to sorafenib was evaluated according to the modified Response Evaluation Criteria in Solid Tumours for Hepatocellular Carcinoma (mRECIST) criteria [23]. Response to therapy was classified as complete response (CR), partial response (PR), stable disease (SD), or progres-sive disease (PD). The disease-control rate (DCR) was defined as the percentage of patients who had a best response rating of complete response, partial response, or stable disease.

The TTP was measured from the starting treatment data until the clinical or radiological evidence of disease progres-sion or last follow-up.

2.4 Statistical Analysis

A two-tailed p-value < 0.05 was considered to indicate statis-tical significance. Categorical data were compared using Fisher’s exact test. Continuous variables were shown as me-dian and compared with the non-parametric Mann–Whitney test. Time-to-event data was visualized using Kaplan–Meier curves and compared by the log-rank test. All statistical anal-yses were performed with software package SPSS for Mac (Rel SPSS 21.0; IBM Corporation, Armonk, NY, USA, 2012).

3 Results

Three hundred thirteen patients with HCC treated with soraf-enib were included in the study. Patients were divided in two groups according to the presence of diabetes: 80 diabetics

Table 1 Baseline characteristics of diabetic and nondiabetic patients Characteristics DIAB n = 80 nDIAB n = 233 P Males 56 (70 %) 175 (75 %) .379

Median age (range) 68 (41–84) 67 (35–90) .805

HCV infection 54 (67.5 %) 152 (65.5 %) .786

HBV infection 9 (11.3 %) 42 (18.0 %) .157

Alcohol 8 (10.0 %) 15 (6.4 %) .292

NAFLD 6 (7.5 %) 11 (4.7 %) .344

Other aetiologies 3 (3.7 %) 13 (5.6 %) .120

Median BMI, (range) 25 (18–45) 25 (18–35) .815

Child score 5 45 (56.3 %) 123 (52.8 %) .145 Child score 6 31 (38.8 %) 80 (34.3 %) Child score 7 4 (5 %) 30 (12.9 %) Ascites 11 (13.8 %) 35 (15 %) .856 Bilirubin≤ 2 mg/dL 76 (95.0 %) 208 (89.3 %) .179 Albumin≥ 3.5 g/dL 57 (71.2 %) 174 (74.7 %) .555 BCLC B 18 (22.5 %) 55 (23.6 %) .880 BCLC C 62 (77.5 %) 178 (76.4 %) Portal thrombosis 41 (51.3 %) 117 (50.2 %) .897 Extrahepatic metastasis 28 (35.0 %) 76 (32.7 %) .783 Median AFP ng/mL (range) 313 (2–433,250) 290 (1–741,720) .690 Previously treated, n 52 (65.0 %) 153 (65.7 %) 1.00

Liver transplantation 1 (1.3 %) 11 (4.7 %) .772

Surgical resection 3 (3.7 %) 5 (2.1 %)

Local ablation 10 (12.5 %) 25 (10.7 %)

TACE 20 (25.0 %) 47 (20.2 %)

Combined locoregional treatments 18 (22.5 %) 65 (27.9 %) No previous treatment 28 (35.0 %) 80 (34.3 %)

Median treatment duration, months 4 (1–41) 3 (1–56) .178 Starting sorafenib dose 800 mg/d 71 (88.8 %) 205 (88.0 %) 1.00

Median daily dose 673 mg 622 mg .496

Median daily dose≥ 400 mg, n of patients 75 (93.8 %) 203 (87.1 %) .148 Median survival, months, (95%CI) 10 (7.6-12.4) 9 (7.5-10.5) .478

(4)

(DIAB) and 233 nondiabetics (nDIAB). No differences in demographics, liver function, and cancer stage between DIAB and nDIAB groups were observed (Table1).

All DIAB patients were affected by type 2 diabetes and the treatment was: exogenous insulin in 57.5 %, oral hypoglyce-mic agents in 28.8 %, acarbose and diet in 13.7 %. After 1–2 cycles of sorafenib, nine (11 %) patients had reduced or

stopped blood glucose-lowering drugs due to the decrease in blood glucose level. One patient experienced severe hypoglycaemia.

The median duration of sorafenib treatment was 4 (range 1–41) months in DIAB and 3 (1–56) months in nDIAB.

Adverse events were observed in 96.3 % and in 94 % of DIAB and nDIAB patients, respectively. Main adverse events (fatigue, hand foot syndrome (HFS), diarrhea, and hyperten-sion) occurred with comparable frequency in both groups (Table2). Rash was the only adverse event more frequent in DIAB as compared to nDIAB: 27.5 % vs 17.6 % (P = .047).

Median sorafenib dose was 621 mg/day (range 55–800) in nDIAB and 673 mg/day (range 81–800) in DIAB group (P = .218). Sorafenib was temporary and permanently discontinued due to adverse events in 23.2 % and 27.2 % in the nDIAB group, and in 21.3 % and 30.4 % in the DIAB group, respectively.

The median overall survival was 9 months (95 % CI, 7.45-10.55) in nDIAB and 10 months (95 % CI, 7.57-12.43) in DIAB group (P = .535) (Fig.1).

Clinical or radiological progression was observed in 107 (45.9 %) patients of the nDIAB group and in 31 (38.8 %) patients of the DIAB group (P = .105). The radiological eval-uation of response after 3 months of therapy was available in 199 (63.6 %) of patients (Table3). The DCR was 71.5 % (103 among 144 patients) and 83.6 % (46 among 55 patients) in the nDIAB and DIAB groups, respectively (P = .10). The proba-bility of disease progression at 6-month and 12-month was 44.0 % and 59.4 % in the nDIAB group, 34.7 % and 44.4 %

Fig. 1 Median overall survival in diabetic and nondiabetic patients

Table 2 Side effects to sorafenib treatment in diabetic and nondiabetic patients

Side effects DIAB Grade 1-2/3-4 nDIAB Grade 1-2/3-4 P Fatigue 48.8 %/3.8 % 45.7 %/3.9 % .973 Anorexia 47.6 %/1.3 % 38.3 %/2.6 % .480 HFS 46.3 %/3.8 % 37.1 %/5.2 % .248 Diarrhea 41.3 %/3.8 % 46.1 %/3.9 % .818 Arterial hypertension 26.3 %/0 % 25.4 %/0.9 % .836 Rash 27.5 %/0 % 15 %/2.6 % .047 Weight loss 22.6 %/0 % 19.8 %/0 % .863 Alopecia 18.9 %/0 % 13.7 %/0 % .682 Pruritus 17.6 %/0 % 11.2 %/0.9 % .372 Abdominal pain 17.5 %/0 % 22.2 %/1.7 % .407 Nausea 15.1 %/2.5 % 14.6 %/0.4 % .427 Jaundice 10.1 %/6.3 % 17.7 %/5.6 % .309 Aftosis 10.1 %/0 % 7.3 %/0.9 % .376 Vomiting 7.6 %/0 % 5.6 %/0.4 % .756 Bleeding 2.5 %/2.5 % 3.9 %/3.3 % .685

(5)

in the DIAB group. Cancer progression was observed in 107 among 233 patients and in 31 among 80, in the nDIAB and DIAB groups, respectively. Median TTP was 19 months (95%CI, 2.06-35.94) in the DIAB group and 9 months (95%CI, 6.51-11.49) in the nDIAB group (P = .038) (Fig.2). In the DIAB group, median TTP was 16 months in insulin-treated patients and 27 months in patients insulin-treated with oral hypoglycemic agents.

4 Discussion

This is the first study that evaluated the interaction between diabetes and sorafenib treatment in a large cohort of HCC patients.

Type 2 diabetes increases the risk of HCC occurrence by two to four fold [6,7]. Liver steatosis and steatohepatitis often observed in diabetics may be a cause for it. However, type 2 diabetes may also stimulate cancer development and progres-sion by hyperglicemia and hyperinsulinemia. According to the

Warburg hypothesis, hyperglicemia favours glycolisis that is a main energy producing process in many cancer cells [24]. Hyperinsulinemia is a risk factor for the development of can-cer [25,26]. Insulin produced by pancreatic cells through portal circulation reaches the liver and, therefore, is exposed to very high concentrations of this hormone [27]. Insulin and insulin-like growth factor receptors are expressed on the sur-face of most cancer cells. Insulin receptor is expressed mainly as A isoform. Both receptors when activated may enhance cancer cell proliferation, metastasis, and inhibit apoptosis. In particular, the activation of A isoform insulin receptors may enhance more cell survival and proliferation than glucose up-take [28]. Another mechanism may be the reduced hepatic synthesis of insulin growth factor-binding protein-1, resulting in increased bioavailability of insulin-like growth factor-1 which leads to cellular proliferation promotion and apoptosis inhibition [29].

The risk of cancer occurrence is higher in patients treated with insulin or sulfonylureas, whereas it is reduced with the use of metformin [8,30,31]. Some epidemiologic studies and basic science suggests an association between metformin use and lower cancer risk in diabetic patients, in particular it is associated with lower incidence and mortality for all cancers. Pathophysiological mechanisms of this significant risk have been proposed, but they are still incompletely defined. Metformin has a potential anti-cancer effect by activating adenosine 5′-mono-phosphate-activated protein kinase (AMPK) in addition to alleviating hyperinsulinemia and hy-perglycemia. [32]. Metformin has also a direct effect on CD8+ tumor-infiltrating lymphocytes protecting them from

Table 3 Radiological response at 3 months in diabetic and nondiabetic patients

Radiological response NDIAB DIAB Progression 41 (28.5 %) 9 (16.4 %)

Stable 65 (45.1 %) 26 (47.3 %)

Partial response 33 (22.9 %) 19 (34.5 %) Complete response 5 (3.5 %) 1 (1.8 %)

Fig. 2 Time-to-progression in diabetic and nondiabetic patients

(6)

apoptosis and counteracting immune exhaustion [33]. Furthermore, metformin seems to inhibit cancer cell prolifer-ation acting as a negative regulator of mTOR [34].

In patients not treated with sorafenib, some studies have shown that diabetes may worsen the outcomes of HCC pa-tients [10,35].

Tyrosine kinase inhibitors can affect the insulin signalling pathway reducing the mitogenic effect and improving glyce-mic control in diabetics. This last effect was observed in 11 % of our patients. Potential mechanisms related to this effect are shown in Table 4. In a single patient a severe episode of hypoglicemia occurred. These findings suggest that patients with diabetes who are treated with sorafenib should be cau-tiously monitored.

The main aim of the study was to test if systemic treatment was as effective and safe in patients with unresectable HCC and type 2 diabetes as compared to patients without diabetes. Sorafenib therapy in diabetic patients was safe. Adverse events occurred with comparable frequency in DIAB and nDIAB patients, except for skin rash that was observed more often in the DIAB group. Median daily dose was comparable between the DIAB and nDIAB groups. Also temporary or definitive discontinuation of sorafenib due to adverse effects was registered with the same rate in the two groups. Overall survival was similar between DIAB and nDIAB groups.

Among secondary outcomes, 3-month radiological re-sponse evaluation showed a lightly higher rate of DCR in DIAB than in nDIAB patients, but the difference did not reach statistical significance.

Unexpectedly, a longer TTP in the DIAB group as com-pared to the nDIAB group (19 vs 9 months, P = .038) was observed. This finding needs to be evaluated in prospective studies. The TTP was longer in DIAB patients treated with oral hypoglycemic agents as compared to those treated with exogenous insulin (27 vs 16 months). These findings contrast with the results of another study showing a shorter progression-free survival and OS in sorafenib-treated patients taking metformin as compared to insulin-treated patients [38]. Definitive conclusions cannot be drawn due to the low num-ber of patients, although previous studies on other cancer types showed that metformin increased the response to che-motherapy and decreased the occurrence of metastasis [39–42]. Furthermore, in a recent study on metastatic renal

carcinoma, for patients treated with sunitinib, the OS was longer in diabetics taking metformin [43].

In conclusion, sorafenib was as safe as effective in DIAB and in nDIAB patients. The longer TTP observed in DIAB as compared to nDIAB patients might suggest a better anticancer effect of sorafenib in patients with diabetes. Also, the better TTP in DIAB treated with oral therapy as compared to those receiving insulin is interesting and concordant with previous studies on other cancer types. However, due to the retrospec-tive design of the analysis, these last findings need to be tested in prospective controlled studies.

Acknowledgments

Author Contributions • Study concept and design: GGDC, RT. • Acquisition of data: RT, GC, LF, LA.

• Analysis and interpretation of data: GGDC, FM, NC. • Drafting of the manuscript: GGDC, MG.

• Critical revision of the manuscript for important intellectual content: GGDC, MG, RT.

• Statistical analysis: GGDC. • Study supervision: GGDC.

All authors read and approved the final manuscript.

Compliance with Ethical Standards Funding None.

Conflict of Interest Giovan Giuseppe Di Costanzo, Raffaella Tortora, Filomena Morisco, Luigi Addario, Maria Guarino, Gabriella Cordone, Luigia Falco, and Nicola Caporaso declare no conflict of interest.

References

1. GLOBOCAN Cancer incidence and mortality worldwide. International Agency for Research on Cancer. World Health Organization, 2012.http://globocan.iarc.fr.

2. Benvegnù L, Gios M, Boccato S, Alberti A. Natural history of compensated viral cirrhosis: a prospective study on the incidence and hierarchy of major complications. Gut. 2004;53:744–9. 3. Sangiovanni A, Prati GM, Fasani P, Ronchi G, Romeo R, Manini

M, et al. The natural history of compensated cirrhosis due to hep-atitis C virus: a 17-year cohort study of 214 patients. Hepatology. 2006;43:1303–10.

4. Ascha MS, Hanouneh IA, Lopez R, Tamimi TAR, Feldstein AF, Zein NN. The incidence and risk factors of hepatocellular carcino-ma in patients with nonalcoholic steatohepatitis. Hepatology. 2010;51:1972–8.

5. Jinjuvadia R, Patel S, Liangpunsakul S. The association between metabolic syndrome and hepatocellular carcinoma: systemic review and meta-analysis. J Clin Gastroenterol. 2014;48(2):172–7. 6. El-Serag HB, Tran T, Everhart JE. Diabetes increases the risk of

c h r o n i c l i v e r d i s e a s e a n d h e p a t o c e l l u l a r c a r c i n o m a . Gastroenterology. 2004;126:460–8.

7. Davila JA, Morgan RO, Shaib Y, McGlynn KA, El-Serag HB. Diabetes increases the risk of hepatocellular carcinoma in the Table 4 Mechanisms potentially involved in diabetes amelioration

during tyrosine kinase inhibitors treatment

Target Effects

VEGF-R capillary regression in pancreatic islets [36] IGF-1, PDGF-R insulin resistance reduction

c-Abl, MAPK JNK, NF-kB decreased apoptosis of beta-cells [37] c-Kit decreased inflammatory response

(7)

United States: a population based case control study. Gut. 2005;54(4):533–9. doi:10.1136/gut.2004.052167.

8. Hassan MM, Curley SA, Li D, Kaseb A, Davila M, Abdalla EK, et al. Association of diabetes duration and diabetes treatment with the risk of hepatocellular carcinoma. Cancer. 2010;116:1938–46. 9. Yuan JM, Govindarajan S, Arakawa K, Yu MC. Synergism of

al-cohol, diabetes and viral hepatitis on the risk of hepatocellular car-cinoma in blacks and whites in the U.S. Cancer. 2004;101:1009–17. 10. Toyoda H, Kumada T, Nakano S, Takeda I, Sugiyama K, Kiriyama S, et al. Impact of diabetes mellitus on the prognosis of patients with hepatocellular carcinoma. Cancer. 2001;91:957–63.

11. Yancik R, Wesley MN, Ries LA, Havlik RJ, Edwards BK, Yates JW. Effect of age and comorbidity in postmenopausal breast cancer patients aged 55 years and older. JAMA. 2001;285:885–92. 12. Meyerhardt JA, Catalano PJ, Haller DG, Mayer RJ, Macdonald JS,

Benson AB, et al. Impact of diabetes mellitus on outcomes in pa-tients with colon cancer. J Clin Oncol. 2003;21:433–40.

13. Coughlin SS, Calle EE, Teras LR, Petrelli J, Thun MJ. Diabetes mellitus as a predictor of cancer mortality in a large cohort of US adults. Am J Epidemiol. 2004;159:1160–7.

14. Bianchi G, Marchesini G, Zoli M, Bugianesi E, Fabbri A, Pisi E. Prognostic significance of diabetes in patients with cirrhosis. Hepatology. 1994;20:119–25.

15. Bruix J, Sherman M. Management of hepatocellular carcinoma. Hepatology. 2005;42:1208–36.

16. Llovet JM, Ricci S, Mazzaferro V, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359(4):378–90. 17. Richardson LC, Pollack LA. Therapy insight: influence of type 2

diabetes on the development, treatment and outcomes of cancer. Nat Clin Pract Oncol. 2005;2:48–53.

18. Van de Poll-Franse LV, Houterman S, Janssen-Heijnen MLG, Dercksen MW, Coebergh JWW, Haak HR. Less aggressive treat-ment and worse overall survival in cancer patients with diabetes: a large population based analysis. Int J Cancer. 2007;120:1986–92. 19. Imarisio I, Paglino C, Ganini C, Magnani L, Caccialanza R, Porta

C. The effect of sorafenib treatment on the diabetic status of patients with renal cell or hepatocellular carcinoma. Future Oncol. 2012;8(8):1051–7.

20. Bruix J, Sherman M, Llovet JM, Beaugrand M, Lencioni R, Burroughs AK, et al. Clinical management of hepatocellular carci-noma. Conclusions of the Barcelona-2000 EASL conference. European Association for the Study of the Liver. J Hepatol. 2001;35:421–30.

21. Llovet JM, Bru C, Bruix J. Prognosis of hepatocellular carci-noma: the BCLC staging classification. Semin Liver Dis. 1999;19:329–38.

22. American Diabetes Association. Classification and diagnosis of di-abetes. Diabetes Care. 2015;38(Suppl):S8–S16.

23. Lencioni R, Llovet JM. Modified RECIST (mRECIST) assessment for hepatocellular carcinoma. Semin Liver Dis. 2010;30:52–60. 24. Vander Heiden MG, Cantley LC, Thompson CB. Understanding

the Warburg effect: the metabolic requirements of cell proliferation. Science. 2009;324:1029–33.

25. Bruning PF, Bonfrer JM, van Noord PA, Hart AA, de Jong-Bakker M, Nooijen WJ. Insulin resistance and breast-cancer risk. Int J Cancer. 1992;52:511–16.

26. Silverman DT, Schiffman M, Everhart J, Goldstein A, Lillemoe KD, Swanson GM, et al. Diabetes mellitus, other medical

conditions and familial history of cancer as risk factors for pancre-atic cancer. Br J Cancer. 1999;80:1830–7.

27. Ferrannini E, Cobelli C. The kinetics of insulin in man. II. Role of the liver. Diabetes Metab Rev. 1987;3:3365–97.

28. Denley A, Carroll JM, Brierley GV, Cosgrove L, Wallace J, Forbes B, et al. Differential activation of insulin receptor substrates 1 and 2 by insulin-like growth factor-activated insulin receptors. Mol Cell Biol. 2007;27:3569–77.

2 9. M o o r e M A , P a r k CB , Tsu d a H . I m p l i c a t i o ns of t h e hyperinsulinaemia-diabetes-cancer link for preventive efforts. Eur J Cancer Prev. 1998;7:89–107.

30. Donadon V, Balbi M, Ghersetti M, Grazioli S, Perciaccante A, Della Valentina G, et al. Antidiabetic therapy and increased risk of hepatocellular carcinoma in chronic liver disease. World J Gastroenterol. 2009;15:2506–11.

31. Bowker SL, Majumdar SR, Veugelers P, Johnson JA. Increased cancer-related mortality for patients with type 2 diabetes who use sulfonylureas or insulin. Diabetes Care. 2006;29:254–8.

32. Noto H, Goto A, Tsujimoto T, Noda M. Cancer risk in diabetic patients treated with metformin: a systematic review and meta-anal-ysis. PLoS One. 2012;7:e33411.

33. Eikawa S, Nishida M, Mizukamia S, Yamazakia C, Nakayamab E, Udonoa H. Immune-mediated antitumor effect by type 2 diabetes drug, metformin. PNAS. 2015;112:1809–14.

34. Sahra IB, Regazzetti C, Robert G, Laurent K, Le Marchand-Brustel Y, Auberger P, et al. Metformin, independent of AMPK, induces mTOR inhibition and cell-cycle arrest through REDD1. Cancer Res. 2011;71:4366–72.

35. Huo T-I, Lui W-Y, Huang Y-H, Chau GY, Wu JC, Lee PC, et al. Diabetes mellitus is a risk factor for hepatic decompensation in patients with hepatocellular carcinoma undergoing resection: a lon-gitudinal study. Am J Gastroenterol. 2003;98:2293–8.

36. Kamba T, Tam BY, Hashizume H, Haskell A, Sennino B, Mancuso MR, et al. VEGF-dependant plasticity of fenestrated capillaries in the normal adult microvasculature. Am J Physiol Heart Circ Physiol. 2006;290:H560–76.

37. Veneri D, Franchini M, Bonora E. Imatinib and regression of type 2 diabetes. N Engl J Med. 2005;10:1049–50.

38. Casadei Gardini A, Marisi G, Scarpi E, Scartozzi M, Faloppi L, Silvestris N, et al. Effects of metformin on clinical outcome in diabetic patients with advanced HCC receiving sorafenib. Expert Opin Pharmacother. 2015;16:2719–25.

39. Li D. Metformin as an antitumor agent in cancer prevention and treatment. J Diabetes. 2001;3(4):320–7.

40. Rocha GZ, Dias MM, Ropelle ER, Osório-Costa F, Rossato FA, Vercesi AE, et al. Metformin amplifies chemotherapy-induced AMPK activation and antitumoral growth. Clin Cancer Res. 2011;17(12):3993–4005.

41. Ben Sahra I, Le Marchand-Brustel Y, Tanti JF, Bost F. Metformin in cancer therapy: a new perspective for an old antidiabetic drug? Mol Cancer Ther. 2010;9(5):1092–9.

42. Rattan R, Ali Fehmi R, Munkarah A. Metformin: an emerging new therapeutic option for targeting cancer stem cells and metastasis. J Oncol. 2012;2012:928127.

43. Keizman D, Ish-Shalom M, Sella A, Gottfried M, Maimon N, Peer A, et al. Metformin use and outcome of sunitinib treatment in pa-tients with diabetes and metastatic renal cell carcinoma. Clin Genitourin Cancer. 2016;S1558–7673(16):30102–1.

Riferimenti

Documenti correlati

In tal senso, implementare il grado di conoscenza di tale processo risulta cruciale al fi ne di aff rontare con successo il governo della complessità: l’ introduzione di un

The purpose of our study was to evaluate safety of ERT and its efficacy on motor, respiratory and cardiac function over the course of long-term treatment in a large cohort of

The computation of the magnetic permeability appears to be harder than the evaluation of the conductivity, but the multi-frequency approach leads to accurate approximations of

Polyphenols are mostly antioxidant and anti-inflammatory compounds, able to modulate abnormal cellular signaling induced by pro-inflammatory stimuli and oxidative stress, as

The maps of the single plant species, either obtained from spatial queries or from modelling, were combined to obtain the following results: total specific richness map (not shown),

In this multifaceted frame of researches, we analysed the alveolar air in a group of patients affected by pancre- atic ductal adenocarcinoma (PDA) cytohistologically confirmed and in

minimo se la regolarizzazione degli errori e delle omissioni avviene entro il termine per la presentazione della dichiarazione relativa all’anno successivo a

– derive a service-controller controlling the number of virtual resources (e.g., VMs or containers) allocated to each network function by using feedback from the true performance of